Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dev Biol ; 477: 241-250, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34052210

RESUMEN

A critical cell type participating in cardiac outflow tract development is a subpopulation of the neural crest cells, the cardiac neural crest cells (NCCs), whose defect causes a spectrum of cardiovascular abnormalities. Accumulating evidence indicates that mTOR, which belongs to the PI3K-related kinase family and impacts multiple signaling pathways in a variety of contexts, plays a pivotal role for NCC development. Here, we investigated functional roles of mTOR for cardiac neural crest development using several lines of mouse genetic models. We found that disruption of mTOR caused NCC defects and failure of cardiac outflow tract separation, which resulted in a spectrum of cardiac defects including persistent truncus arteriosus, ventricular septal defect and ventricular wall defect. Specifically, mutant neural crest cells showed reduced migration into the cardiac OFT and prematurely exited the cell cycle. A number of critical factors and fundamental signaling pathways, which are important for neural crest and cardiomyocyte development, were impaired. Moreover, actin dynamics was disrupted by mTOR deletion. Finally, by phenotyping the neural crest Rptor and Rictor knockout mice respectively, we demonstrate that mTOR acts principally through the mTORC1 pathway for cardiac neural crest cells. Altogether, these data established essential roles of mTOR for cardiac NCC development and imply that dysregulation of mTOR in NCCs may underline a spectrum of cardiac defects.


Asunto(s)
Anomalías Cardiovasculares/genética , Corazón/embriología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Miocardio/metabolismo , Cresta Neural/embriología , Serina-Treonina Quinasas TOR/fisiología , Animales , Células Cultivadas , Eliminación de Gen , Redes y Vías Metabólicas , Ratones , Cresta Neural/metabolismo , Serina-Treonina Quinasas TOR/genética
2.
Dev Biol ; 467(1-2): 77-87, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32866472

RESUMEN

We herein report that deletion of mTOR in dental epithelia caused defective development of multiple cell layers of the enamel organ, which culminated in tooth malformation and cystogenesis. Specifically, cells of the stellate reticulum and stratum intermedium were poorly formed, resulting in cystic changes. The pre-ameloblasts failed to elongate along the apical-basal axis and persisted vigorous expression of Sox2 and P63, which are normally downregulated during cytodifferentiation. Expression of amelogenic markers was also attenuated in mutants. Cell proliferation and cell sizes in mutants were significantly reduced over time. Importantly, we found reduced amounts and aberrant aggregations of cytoskeletal components in mutants, along with attenuated expression of cytoskeleton regulator Cdc42, whose epithelial deletion causes a similar phenotype. Moreover, disruption of actin assembly in an organ culture system affected cell proliferation and cytodifferentiation of tooth germs, supporting a causative role of mTOR-regulated cytoskeleton dynamics for the observed phenotype of mTOR mutant mice. In further support of this view, we showed that mTOR overactivation caused increased cytoskeletal component synthesis and assembly, along with accelerated cytodifferentiation in the enamel organ. Finally, we demonstrated that mTOR regulated enamel organ development principally through the mTORC1 pathway.


Asunto(s)
Citoesqueleto/metabolismo , Órgano del Esmalte/embriología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Citoesqueleto/genética , Órgano del Esmalte/citología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Transgénicos , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Serina-Treonina Quinasas TOR/genética , Transactivadores/genética , Transactivadores/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
3.
PLoS Genet ; 14(7): e1007491, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29975682

RESUMEN

mTOR is a highly conserved serine/threonine protein kinase that is critical for diverse cellular processes in both developmental and physiological settings. mTOR interacts with a set of molecules including Raptor and Rictor to form two distinct functional complexes, namely the mTORC1 and mTORC2. Here, we used novel genetic models to investigate functions of the mTOR pathway for cranial neural crest cells (NCCs), which are a temporary type of cells arising from the ectoderm layer and migrate to the pharyngeal arches participating craniofacial development. mTOR deletion elicited a proliferation deficit and excessive apoptosis of post-migratory NCCs, leading to growth arrest of the facial primordia along with midline orofacial clefts. Furthermore, NCC differentiation was impaired. Thus, NCC derivatives, such as skeletons, vasculatures and neural tissues were either rudimentary or malformed. We further demonstrate that disruption of mTOR caused P53 hyperactivity and cell cycle arrest in cranial NCCs, and lowering P53 activity by one copy reduction attenuated the severity of craniofacial phenotype in NCC-mTOR knockout mice. Remarkably, NCC-Rptor disruption caused a spectrum of defects mirroring that of the NCC-mTOR deletion, whereas NCC-Rictor disruption only caused a mild craniofacial phenotype compared to the mTOR and Rptor conditional knockout models. Altogether, our data demonstrate that mTOR functions mediated by mTORC1 are indispensable for multiple processes of NCC development including proliferation, survival, and differentiation during craniofacial morphogenesis and organogenesis, and P53 hyperactivity in part accounts for the defective craniofacial development in NCC-mTOR knockout mice.


Asunto(s)
Anomalías Craneofaciales/genética , Cresta Neural/embriología , Transducción de Señal/fisiología , Cráneo/embriología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/fisiología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Anomalías Craneofaciales/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Morfogénesis/fisiología , Cresta Neural/citología , Cresta Neural/metabolismo , Organogénesis/fisiología , Serina-Treonina Quinasas TOR/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Stem Cells ; 33(8): 2483-95, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26010009

RESUMEN

Adipogenesis is essential for soft tissue reconstruction following trauma or tumor resection. We demonstrate that CD31(-)/34(+)/146(-) cells, a subpopulation of the stromal vascular fraction (SVF) of human adipose tissue, were robustly adipogenic. Insulin growth factor-1 (IGF1) promoted a lineage bias towards CD31(-)/34(+)/146(-) cells at the expense of CD31(-)/34(+)/146(+) cells. IGF1 was microencapsulated in poly(lactic-co-glycolic acid) scaffolds and implanted in the inguinal fat pad of C57Bl6 mice. Control-released IGF1 induced remarkable adipogenesis in vivo by recruiting endogenous cells. In comparison with the CD31(-)/34(+)/146(+) cells, CD31(-)/34(+)/146(-) cells had a weaker Wnt/ß-catenin signal. IGF1 attenuated Wnt/ß-catenin signaling by activating Axin2/PPARγ pathways in SVF cells, suggesting IGF1 promotes CD31(-)/34(+)/146(-) bias through tuning Wnt signal. PPARγ response element (PPRE) in Axin2 promoter was crucial for Axin2 upregulation, suggesting that PPARγ transcriptionally activates Axin2. Together, these findings illustrate an Axin2/PPARγ axis in adipogenesis that is particularly attributable to a lineage bias towards CD31(-)/34(+)/146(-) cells, with implications in adipose regeneration.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Antígenos de Diferenciación/metabolismo , Diferenciación Celular , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre/metabolismo , Adulto , Animales , Femenino , Humanos , Ratones , Persona de Mediana Edad
5.
FASEB J ; 29(10): 4083-91, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26148970

RESUMEN

As transcription factors of the lines (LIN)-11/Islet (Isl)-1/mitosis entry checkpoint (MEC)-3 (LIM)-homeobox subfamily, LIM homeobox (Lhx)6 and -8 are remarkably conserved and involved in the morphogenesis of multiple organ systems. Lhx6 and -8 play overlapping and distinctive roles, but in general act as cell fate mediators and in turn are regulated by several transcriptional factors, such as sonic hedgehog, fibroblast growth factors, and wingless-int (Wnt)/ß-catenin. In this review, we first summarize Lhx6 and -8 distributions in development and then explore how Lhx6 and -8 act as transcription factors and coregulators of cell lineage specification. Known Lhx6 and -8 functions and targets are outlined in neurogenesis, craniofacial development, and germ cell differentiation. The underlying mechanisms of Lhx6 and -8 in regulating cell fate remain elusive. Whether Lhx6 and -8 affect functions in tissues and organs other than neural, craniofacial, oocytes, and germ cells is largely unexplored. Taken together, Lhx6 and -8 are important regulators of cell lineage specification and may act as one of the pivotal mediators of stem cell fate. Undoubtedly, future investigations of Lhx6 and -8 biology will continue to yield fascinating insights into tissue development and homeostasis, in addition to their putative roles in tissue regeneration and ageing.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Diente/metabolismo , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Humanos , Proteínas con Homeodominio LIM/metabolismo , Ratones , Modelos Genéticos , Proteínas del Tejido Nervioso/metabolismo , Diente/embriología , Factores de Transcripción/metabolismo
6.
Mater Today Bio ; 24: 100933, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38283982

RESUMEN

Injured articular cartilage is a leading cause for osteoarthritis. We recently discovered that endogenous stem/progenitor cells not only reside in the superficial zone of mouse articular cartilage, but also regenerated heterotopic bone and cartilage in vivo. However, whether critical-size osteochondral defects can be repaired by pure induced chemotatic cell homing of these endogenous stem/progenitor cells remains elusive. Here, we first found that cells in the superficial zone of articular cartilage surrounding surgically created 3 × 1 mm defects in explant culture of adult goat and rabbit knee joints migrated into defect-filled fibrin/hylaro1nate gel, and this migration was significantly more robust upon delivery of exogenous granulocyte-colony stimulating factor (G-CSF). Remarkably, G-CSF-recruited chondrogenic progenitor cells (CPCs) showed significantly stronger migration ability than donor-matched chondrocytes and osteoblasts. G-CSF-recruited CPCs robustly differentiated into chondrocytes, modestly into osteoblasts, and barely into adipocytes. In vivo, critical-size osteochondral defects were repaired by G-CSF-recruited endogenous cells postoperatively at 6 and 12 weeks in comparison to poor healing by gel-only group or defect-only group. ICRS and O'Driscoll scores of articular cartilage were significantly higher for both 6- and 12-week G-CSF samples than corresponding gel-only and defect-only groups. Thus, endogenous stem/progenitor cells may be activated by G-CSF, a Food and Drug Administration (FDA)-cleared bone-marrow stimulating factor, to repair osteochondral defects.

7.
Microsurgery ; 33(5): 367-75, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23653396

RESUMEN

Adipose tissue-derived stem cells and insulin-like growth factor-1 (IGF-1) have shown potential to enhance peripheral nerve regeneration. The purpose of this study was to investigate the effect of an in vivo biologic scaffold, consisting of white adipose tissue flap (WATF) and/or IGF-1 on nerve regeneration in a crush injury model. Forty rats all underwent a sciatic nerve crush injury and then received: a pedicled WATF, a controlled local release of IGF-1, both treatments, or no treatment at the injury site. Outcomes were the normalized maximum isometric tetanic force (ITF) of the tibialis anterior muscle and histomorphometric measurements. At 4 weeks, groups with WATF had a statistically significant improvement in maximum ITF recovery, as compared to those without (P < 0.05), and there was an increase in myelin thickness and total axon count in the WATF-only group versus control (P < 0.01). Functional and histomorphometric data suggest that IGF-1 suppressed the effect of the WATF. Use of a pedicled WATF improved the functional and histomorphometrical results after axonotmesis in a rat model. IGF-1 does not appear to enhance nerve regeneration in this model. Utilizing the WATF may have a beneficial therapeutic role in peripheral nerve injuries.


Asunto(s)
Tejido Adiposo Blanco , Regeneración Tisular Dirigida/métodos , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/terapia , Nervio Ciático/lesiones , Colgajos Quirúrgicos , Animales , Fenómenos Biomecánicos , Terapia Combinada , Preparaciones de Acción Retardada , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Microesferas , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Traumatismos de los Nervios Periféricos/cirugía , Ratas , Ratas Sprague-Dawley , Nervio Ciático/efectos de los fármacos , Nervio Ciático/patología , Nervio Ciático/fisiología , Andamios del Tejido , Resultado del Tratamiento
8.
J Cell Biochem ; 113(9): 2903-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22531959

RESUMEN

Upregulation of miR-34a by p53 is recently believed to be a key mediator in the pro-apoptotic effects of this tumor suppressor. We sought to determine whether restoration of miR-34a levels in p53 deficient cells could rescue the response to DNA damage. Compared with the p53 wildtype U2OS cells, miR-34a expression was much lower in p53 deficient Saos2 cells upon cisplatin treatment. Unexpectedly, delivery of miR-34a in Saos2 cells does not increase the cell sensitivity to apoptosis. This effect was mediated by direct downregulation of SirT1 expression by miR-34a, which in turn increased the NFκB activity. Inhibition of NFκB activity in Saos2 cells by Aspirin sensitized the miR-34a overexpressing cells to cell death. Thus, in tumors with p53 deficiency, miR-34a restoration alone confers drug resistance through Sirt1-NFκB pathway and combination of miR-34a and NFκB inhibitor could be considered as a promising therapeutic strategy.


Asunto(s)
MicroARNs/metabolismo , FN-kappa B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Humanos , MicroARNs/genética , FN-kappa B/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética
9.
Cell Tissue Res ; 347(3): 665-76, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22382390

RESUMEN

From its inception, tissue engineering has had three tenets: cells, biomaterial scaffolds and signaling molecules. Among the triad, cells are the center piece, because cells are the building blocks of tissues. For decades, cell therapies have focused on the procurement, manipulation and delivery of healthy cells for the treatment of diseases or trauma. Given the complexity and potential high cost of cell delivery, there is recent and surging interest to orchestrate endogenous cells for tissue regeneration. Biomaterial scaffolds are vital for many but not all, tissue-engineering applications and serve to accommodate or promote multiple cellular functions. Signaling molecules can be produced by transplanted cells or endogenous cells, or delivered specifically to regulate cell functions. This review highlights recent work in tissue engineering and cell therapies, with a focus on harnessing the capacity of endogenous cells as an alternative or adjunctive approach for tissue regeneration.


Asunto(s)
Fenómenos Fisiológicos Musculoesqueléticos , Células Madre/citología , Ingeniería de Tejidos/métodos , Animales , Humanos , Trasplante de Células Madre
10.
FASEB J ; 25(10): 3496-504, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21746864

RESUMEN

Cell transplantation has been well explored for cartilage regeneration. We recently showed that the entire articular surface of a synovial joint can regenerate by endogenous cell homing and without cell transplantation. However, the sources of endogenous cells that regenerate articular cartilage remain elusive. Here, we studied whether cytokines not only chemotactically recruit adipose stem cells (ASCs), mesenchymal stem cells (MSCs), and synovium stem cells (SSCs) but also induce chondrogenesis of the recruited cells. Recombinant human transforming growth factor-ß3 (TGF-ß3; 100 ng) and/or recombinant human stromal derived factor-1ß (SDF-1ß; 100 ng) was control released into an acellular collagen sponge cube with underlying ASCs, MSCs, or SSCs in monolayer culture. Although all cell types randomly migrated into the acellular collagen sponge cube, TGF-ß3 and/or SDF-1ß recruited significantly more cells than the cytokine-free control group. In 6 wk, TGF-ß3 alone recruited substantial numbers of ASCs (558±65) and MSCs (302±52), whereas codelivery of TGF-ß3 and SDF-1ß was particularly chemotactic to SSCs (400±120). Proliferation of the recruited cells accounted for some, but far from all, of the observed cellularity. TGF-ß3 and SDF-1ß codelivery induced significantly higher aggrecan gene expression than the cytokine-free group for ASCs, MSCs, and SSCs. Type II collagen gene expression was also significantly higher for ASCs and SSCs by SDF-1 and TGF-ß3 codelivery. Remarkably, the expression of aggrecan and type II collagen was detected among all cell types. Thus, homing of multiple stem/progenitor cell populations may potentially serve as an alternative or adjunctive approach to cell transplantation for cartilage regeneration.


Asunto(s)
Adipocitos/fisiología , Células de la Médula Ósea/fisiología , Quimiotaxis/fisiología , Condrogénesis/fisiología , Células Madre/fisiología , Membrana Sinovial/citología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Citocinas , Humanos , Células Madre/efectos de los fármacos , Membrana Sinovial/fisiología , Ingeniería de Tejidos , Andamios del Tejido , Factor de Crecimiento Transformador beta3/farmacología
11.
Bioact Mater ; 15: 316-329, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35356816

RESUMEN

Utilization of the body's regenerative potential for tissue repair is known as in situ tissue regeneration. However, the use of exogenous growth factors requires delicate control of the dose and delivery strategies and may be accompanied by safety, efficacy and cost concerns. In this study, we developed, for the first time, a biomaterial-based strategy to activate endogenous transforming growth factor beta 1 (TGFß1) under alkaline conditions for effective in situ tissue regeneration. We demonstrated that alkaline-activated TGFß1 from blood serum, bone marrow fluids and soaking solutions of meniscus and tooth dentin was capable of increasing cell recruitment and early differentiation, implying its broad practicability. Furthermore, we engineered an injectable hydrogel (MS-Gel) consisting of gelatin microspheres for loading strong alkaline substances and a modified gelatin matrix for hydrogel click crosslinking. In vitro models showed that alkaline MS-Gel controllably and sustainably activated endogenous TGFß1 from tooth dentin for robust bone marrow stem cell migration. More importantly, infusion of in vivo porcine prepared root canals with alkaline MS-Gel promoted significant pulp-dentin regeneration with neurovascular stroma and mineralized tissue by endogenous proliferative cells. Therefore, this work offers a new bench-to-beside translation strategy using biomaterial-activated endogenous biomolecules to achieve in situ tissue regeneration without the need for cell or protein delivery.

12.
Lancet ; 376(9739): 440-8, 2010 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-20692530

RESUMEN

BACKGROUND: A common approach for tissue regeneration is cell delivery, for example by direct transplantation of stem or progenitor cells. An alternative, by recruitment of endogenous cells, needs experimental evidence. We tested the hypothesis that the articular surface of the synovial joint can regenerate with a biological cue spatially embedded in an anatomically correct bioscaffold. METHODS: In this proof of concept study, the surface morphology of a rabbit proximal humeral joint was captured with laser scanning and reconstructed by computer-aided design. We fabricated an anatomically correct bioscaffold using a composite of poly-epsilon-caprolactone and hydroxyapatite. The entire articular surface of unilateral proximal humeral condyles of skeletally mature rabbits was surgically excised and replaced with bioscaffolds spatially infused with transforming growth factor beta3 (TGFbeta3)-adsorbed or TGFbeta3-free collagen hydrogel. Locomotion and weightbearing were assessed 1-2, 3-4, and 5-8 weeks after surgery. At 4 months, regenerated cartilage samples were retrieved from in vivo and assessed for surface fissure, thickness, density, chondrocyte numbers, collagen type II and aggrecan, and mechanical properties. FINDINGS: Ten rabbits received TGFbeta3-infused bioscaffolds, ten received TGFbeta3-free bioscaffolds, and three rabbits underwent humeral-head excision without bioscaffold replacement. All animals in the TGFbeta3-delivery group fully resumed weightbearing and locomotion 3-4 weeks after surgery, more consistently than those in the TGFbeta3-free group. Defect-only rabbits limped at all times. 4 months after surgery, TGFbeta3-infused bioscaffolds were fully covered with hyaline cartilage in the articular surface. TGFbeta3-free bioscaffolds had only isolated cartilage formation, and no cartilage formation occurred in defect-only rabbits. TGFbeta3 delivery yielded uniformly distributed chondrocytes in a matrix with collagen type II and aggrecan and had significantly greater thickness (p=0.044) and density (p<0.0001) than did cartilage formed without TGFbeta3. Compressive and shear properties of TGFbeta3-mediated articular cartilage did not differ from those of native articular cartilage, and were significantly greater than those of cartilage formed without TGFbeta3. Regenerated cartilage was avascular and integrated with regenerated subchondral bone that had well defined blood vessels. TGFbeta3 delivery recruited roughly 130% more cells in the regenerated articular cartilage than did spontaneous cell migration without TGFbeta3. INTERPRETATION: Our findings suggest that the entire articular surface of the synovial joint can regenerate without cell transplantation. Regeneration of complex tissues is probable by homing of endogenous cells, as exemplified by stratified avascular cartilage and vascularised bone. Whether cell homing acts as an adjunctive or alternative approach of cell delivery for regeneration of tissues with different organisational complexity warrants further investigation. FUNDING: New York State Stem Cell Science; US National Institutes of Health.


Asunto(s)
Regeneración Ósea , Cartílago Articular/fisiología , Regeneración , Ingeniería de Tejidos , Factor de Crecimiento Transformador beta3/farmacología , Animales , Movimiento Celular , Conejos , Articulación del Hombro , Andamios del Tejido
13.
Am J Dent ; 24(6): 331-5, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22263328

RESUMEN

PURPOSE: To explore a relationship between the size of pulp chamber perforation and reparative dentin formation in a canine model. METHODS: Pulp defects were created in the pulp chambers of maxillary and mandibular premolars (N = 64) in 17 healthy mongrel dogs in three different sizes (diameter/depth: 1/1, 2/1, and 2/2 mm3) with sterile round burs under general anesthesia. The perforations were immediately capped with hard-setting calcium hydroxide (CH) in the control group or sealed with Teflon membrane (TM) in the experimental group, followed by restoration with reinforced zinc oxide eugenol cement in vivo. Seven and 30 days after pulp chamber perforation and restoration all treated and control premolars were extracted and prepared for histomorphometric and statistical analyses. RESULTS: Reparative dentin formation was more pronounced for defect sizes up to 2/1 mm3 when treated with CH, and completely bridged the surgically created dentin defects only after 30 days. However, reparative dentin upon CH treatment failed to completely bridge pulp chamber exposure for 2/2 defects. By contrast, TM treatment only yielded mild reparative dentin bridging for defects up to 1/1, but not for either 2/1 or 2/2 defects at 30 days. Inflammatory responses of the exposed dental pulp tissue were more robust with the TM group than with the CH group. Thus, dental pulp tissue possesses a capacity for spontaneous repair by the formation of reparative dentin in this preclinical model, but only up to a defect size of -2 mm in diameter and 1 mm in depth. All observations are based on 30 days post-treatment in the canine model. These findings may serve as baseline for regenerative endodontic studies.


Asunto(s)
Recubrimiento de la Pulpa Dental/veterinaria , Pulpa Dental/patología , Modelos Animales de Enfermedad , Animales , Perros
14.
Methods ; 47(2): 116-21, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18952179

RESUMEN

Tissue engineering offers the possibility for soft tissue reconstruction and augmentation without autologous grafting or conventional synthetic materials. Two critical challenges have been addressed in a number of recent studies: a biology challenge of angiogenesis and an engineering challenge of shape maintenance. These two challenges are inter-related and are effectively addressed by integrated bioengineering strategies. Recently, several integrated bioengineering strategies have been applied to improve bioengineered adipose tissue grafts, including internalized microchannels, delivery of angiogenic growth factors, tailored biomaterials and transplantation of precursor cells with continuing differentiation potential. Bioengineered soft tissue grafts are only clinically meaningful if they are vascularized, maintain shape and dimensions, and remodel with the host. Ongoing studies have begun to demonstrate the feasibility towards an ultimate goal to generate vascularized soft tissue grafts that maintain anatomically desirable shape and dimensions.


Asunto(s)
Tejido Adiposo , Ingeniería de Tejidos/métodos , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/trasplante , Ingeniería Biomédica , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Humanos , Hidrogeles/uso terapéutico , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Polietilenglicoles/uso terapéutico , Procedimientos de Cirugía Plástica/métodos , Andamios del Tejido , Trasplante Autólogo
15.
Facial Plast Surg ; 26(5): 413-20, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20853233

RESUMEN

Current autologous fat grafting technique suffers from the drawbacks of donor site morbidity and, more importantly, significant resorption of the grafted fat. Adipose tissue engineering using adult human stem cells has been found to overcome the shortcomings of autologous fat grafting in reconstructing facial defects. Mesenchymal stem cells that can self-renew and differentiate into mature adipocytes have been used to generate adipose tissue, in both in vitro and in vivo cell transplantation studies. However, long-term maintenance of the shape and dimension of the produced adipose tissue remains a challenge, even in tissue engineering with cell transplantation. The choice of appropriate scaffolds to promote stem cell adhesion, proliferation, and differentiation is essential for successful adipogenesis. Recent advances in nanotechnology allow the development of nanostructured scaffolds with a cellular environment that maximally enhances not only cell expansion but also the neovascularization that is crucial for long-term maintenance of cell volume. Cell homing is a technique that actively recruits endogenous host stem cells into a predefined anatomic location for the desired tissue generation. Bypassing ex vivo cell manipulation, the cell homing technique eliminates donor site morbidity and rejection, reducing the regulation issue in clinical translation. Mao et al. introduced the concept of biosurgery, which combined nanostructured scaffolds and growth factor biocues, with or without cell transplantation, for successful de novo adipogenesis in restoring facial defects. Important questions, such as the necessity of cell transplantation in scaling up the size of engineered adipose tissue, need to be answered with further studies. However, the era of biosurgery replacing conventional treatments such as biologically inactive filler injections and alloplastic implants appears to be in the near future.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/citología , Células Madre Adultas/citología , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/métodos , Adipocitos/fisiología , Adulto , Células Madre Adultas/fisiología , Biotecnología , Técnicas de Cultivo de Célula , Humanos
16.
Semin Orthod ; 16(2): 143-146, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25018618

RESUMEN

The profession of orthodontics is projected to face a multitude of challenges. Do cyclic forces accelerate the rate of tooth movement and hence the speed of orthodontic treatment? Would bioengineered cementum and dentine be a solution to root resorption? What would orthodontics be like when bioengineered periodontal ligament and alveolar bone become clinical practice, or one day, entire teeth are bioengineered? Would it be possible to selectively differentiate stem cells into osteoblasts or osteoclasts by either static or cyclic forces? What is the new demand on orthodontic expertise with increasingly automated appliances? What will be the impact of the next generation of dental implants or rapid prototyped crowns on orthodontics? A century ago, Edward Angle's practice of fixed appliances, along with other seminal contributions, such as functional appliances, established the profession of orthodontics. Today, the biophysical principles of orthodontics remain largely unchanged from Angle's era, despite incremental refinements of brackets and wires. The paucity of fundamental innovations in orthodontics for decades presents intrinsic risks for the profession. This review will identify challenges for contemporary orthodontics and delineate strategies for the profession to evolve in an era of unprecedented scientific and technological advances, and serve as a call to action for the orthodontic profession.

17.
J Orthop Res ; 38(3): 485-502, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31994782

RESUMEN

Cell-based therapies, defined here as the delivery of cells in vivo to treat disease, have recently gained increasing public attention as a potentially promising approach to restore structure and function to musculoskeletal tissues. Although cell-based therapy has the potential to improve the treatment of disorders of the musculoskeletal system, there is also the possibility of misuse and misrepresentation of the efficacy of such treatments. The medical literature contains anecdotal reports and research studies, along with web-based marketing and patient testimonials supporting cell-based therapy. Both the American Society for Bone and Mineral Research (ASBMR) and the Orthopaedic Research Society (ORS) are committed to ensuring that the potential of cell-based therapies is realized through rigorous, reproducible, and clinically meaningful scientific discovery. The two organizations convened a multidisciplinary and international Task Force composed of physicians, surgeons, and scientists who are recognized experts in the development and use of cell-based therapies. The Task Force was charged with defining the state-of-the art in cell-based therapies and identifying the gaps in knowledge and methodologies that should guide the research agenda. The efforts of this Task Force are designed to provide researchers and clinicians with a better understanding of the current state of the science and research needed to advance the study and use of cell-based therapies for skeletal tissues. The design and implementation of rigorous, thorough protocols will be critical to leveraging these innovative treatments and optimizing clinical and functional patient outcomes. In addition to providing specific recommendations and ethical considerations for preclinical and clinical investigations, this report concludes with an outline to address knowledge gaps in how to determine the cell autonomous and nonautonomous effects of a donor population used for bone regeneration. © 2020 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:485-502, 2020.

18.
J Bone Miner Res ; 35(1): 3-17, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31545883

RESUMEN

Cell-based therapies, defined here as the delivery of cells in vivo to treat disease, have recently gained increasing public attention as a potentially promising approach to restore structure and function to musculoskeletal tissues. Although cell-based therapy has the potential to improve the treatment of disorders of the musculoskeletal system, there is also the possibility of misuse and misrepresentation of the efficacy of such treatments. The medical literature contains anecdotal reports and research studies, along with web-based marketing and patient testimonials supporting cell-based therapy. Both the American Society for Bone and Mineral Research (ASBMR) and the Orthopaedic Research Society (ORS) are committed to ensuring that the potential of cell-based therapies is realized through rigorous, reproducible, and clinically meaningful scientific discovery. The two organizations convened a multidisciplinary and international Task Force composed of physicians, surgeons, and scientists who are recognized experts in the development and use of cell-based therapies. The Task Force was charged with defining the state-of-the art in cell-based therapies and identifying the gaps in knowledge and methodologies that should guide the research agenda. The efforts of this Task Force are designed to provide researchers and clinicians with a better understanding of the current state of the science and research needed to advance the study and use of cell-based therapies for skeletal tissues. The design and implementation of rigorous, thorough protocols will be critical to leveraging these innovative treatments and optimizing clinical and functional patient outcomes. In addition to providing specific recommendations and ethical considerations for preclinical and clinical investigations, this report concludes with an outline to address knowledge gaps in how to determine the cell autonomous and nonautonomous effects of a donor population used for bone regeneration. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Ortopedia , Comités Consultivos , Huesos , Humanos , Minerales , Sociedades Médicas , Estados Unidos
19.
FASEB J ; 22(6): 1684-93, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18198217

RESUMEN

The common premise of synthetic implants in the restoration of diseased tissues and organs is to use inert and solid materials. Here, a porous titanium implant was fabricated for the delivery of microencapsulated bioactive cues. Control-released transforming growth factor-beta1 (TGF-beta1) promoted the proliferation and migration of human mesenchymal stem cells into porous implants in vitro. At 4 wk of implantation in the rabbit humerus, control-released TGF-beta1 from porous implants significantly increased bone-to-implant contact (BIC) by 96% and bone ingrowth by 50% over placebos. Control-released 100 ng TGF-beta1 induced equivalent BIC and bone ingrowth to adsorbed 1 microg TGF-beta1, suggesting that controlled release is effective at 10-fold less drug dose than adsorption. Histomorphometry, scanning electron microscopy, and microcomputed tomography showed that control-released TGF-beta1 enhanced bone ingrowth in the implant's pores and surface. These findings suggest that solid prostheses can be transformed into porous implants to serve as drug delivery carriers, from which control-released bioactive cues augment host tissue integration.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Oseointegración/efectos de los fármacos , Prótesis e Implantes , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Húmero , Células Madre Mesenquimatosas/citología , Porosidad , Conejos , Titanio , Factor de Crecimiento Transformador beta1/administración & dosificación
20.
Am J Transl Res ; 11(3): 1736-1747, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30972197

RESUMEN

Temporomandibular joint (TMJ) arthritis causes severe debilitation and has few treatment options. Here, we found a small molecule, DNA methyltransferase 3B (Dnmt3b), as a putative therapeutic target, partially rescued osteoarthritic phenotype. Dnmt3b was detected differentially expressed in cell zones of mandibular condylar cartilage and the expression of Dnmt3b decreased in the progression of TMJ osteoarthritis. Dnmt3b deficiency using conditional knockout mice led to the onset of osteoarthritis-like conditions including cartilage clefts, cartilage matrix loss and premature chondrocyte hypertrophy, which suggested that Dnmt3b functioned as a osteoarthritis suppressor. Dnmt3b gain-of-function in TMJ stem/progenitor cells showed increases in collagen type II but decreases in collagen type X, whereas Dnmt3b knockdown had opposite effects with attenuated collagen type II but increased collagen type X. Dnmt3b acted via Wnt/ß-catenin signaling and Dnmt3b regulated TMJ stem/progenitor cells differentiation by inducing their premature progression towards hypertrophic chondrocytes through ß-catenin transnucleation and activation. Finally, local Dnmt3b delivery partially rescued cartilage degradation in experimentally induced osteoarthritis. Thus, novel molecules in articular cartilage, such as Dnmt3b, may have therapeutic effects for TMJ osteoarthritis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA